1
|
Zhang H, Wang J, Qu Y, Yang Y, Guo ZN. Brain injury biomarkers and applications in neurological diseases. Chin Med J (Engl) 2025; 138:5-14. [PMID: 38915214 PMCID: PMC11717530 DOI: 10.1097/cm9.0000000000003061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Indexed: 06/26/2024] Open
Abstract
ABSTRACT Neurological diseases are a major health concern, and brain injury is a typical pathological process in various neurological disorders. Different biomarkers in the blood or the cerebrospinal fluid are associated with specific physiological and pathological processes. They are vital in identifying, diagnosing, and treating brain injuries. In this review, we described biomarkers for neuronal cell body injury (neuron-specific enolase, ubiquitin C-terminal hydrolase-L1, αII-spectrin), axonal injury (neurofilament proteins, tau), astrocyte injury (S100β, glial fibrillary acidic protein), demyelination (myelin basic protein), autoantibodies, and other emerging biomarkers (extracellular vesicles, microRNAs). We aimed to summarize the applications of these biomarkers and their related interests and limits in the diagnosis and prognosis for neurological diseases, including traumatic brain injury, status epilepticus, stroke, Alzheimer's disease, and infection. In addition, a reasonable outlook for brain injury biomarkers as ideal detection tools for neurological diseases is presented.
Collapse
Affiliation(s)
- Han Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jing Wang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
- Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
2
|
Gruol DL. The Neuroimmune System and the Cerebellum. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2511-2537. [PMID: 37950146 PMCID: PMC11585519 DOI: 10.1007/s12311-023-01624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
The recognition that there is an innate immune system of the brain, referred to as the neuroimmune system, that preforms many functions comparable to that of the peripheral immune system is a relatively new concept and much is yet to be learned. The main cellular components of the neuroimmune system are the glial cells of the brain, primarily microglia and astrocytes. These cell types preform many functions through secretion of signaling factors initially known as immune factors but referred to as neuroimmune factors when produced by cells of the brain. The immune functions of glial cells play critical roles in the healthy brain to maintain homeostasis that is essential for normal brain function, to establish cytoarchitecture of the brain during development, and, in pathological conditions, to minimize the detrimental effects of disease and injury and promote repair of brain structure and function. However, dysregulation of this system can occur resulting in actions that exacerbate or perpetuate the detrimental effects of disease or injury. The neuroimmune system extends throughout all brain regions, but attention to the cerebellar system has lagged that of other brain regions and information is limited on this topic. This article is meant to provide a brief introduction to the cellular and molecular components of the brain immune system, its functions, and what is known about its role in the cerebellum. The majority of this information comes from studies of animal models and pathological conditions, where upregulation of the system facilitates investigation of its actions.
Collapse
Affiliation(s)
- Donna L Gruol
- Neuroscience Department, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
3
|
Di Mauro G, González VJ, Bambini F, Camarda S, Prado E, Holgado JP, Vázquez E, Ballerini L, Cellot G. MoS 2 2D materials induce spinal cord neuroinflammation and neurotoxicity affecting locomotor performance in zebrafish. NANOSCALE HORIZONS 2024; 9:785-798. [PMID: 38466179 DOI: 10.1039/d4nh00041b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
MoS2 nanosheets belong to an emerging family of nanomaterials named bidimensional transition metal dichalcogenides (2D TMDCs). The use of such promising materials, featuring outstanding chemical and physical properties, is expected to increase in several fields of science and technology, with an enhanced risk of environmental dispersion and associated wildlife and human exposures. In this framework, the assessment of MoS2 nanosheets toxicity is instrumental to safe industrial developments. Currently, the impact of the nanomaterial on the nervous tissue is unexplored. In this work, we use as in vivo experimental model the early-stage zebrafish, to investigate whether mechano-chemically exfoliated MoS2 nanosheets reach and affect, when added in the behavioral ambient, the nervous system. By high throughput screening of zebrafish larvae locomotor behavioral changes upon exposure to MoS2 nanosheets and whole organism live imaging of spinal neuronal and glial cell calcium activity, we report that sub-acute and prolonged ambient exposures to MoS2 nanosheets elicit locomotor abnormalities, dependent on dose and observation time. While 25 μg mL-1 concentration treatments exerted transient effects, 50 μg mL-1 ones induced long-lasting changes, correlated to neuroinflammation-driven alterations in the spinal cord, such as astrogliosis, glial intracellular calcium dysregulation, neuronal hyperactivity and motor axons retraction. By combining integrated technological approaches to zebrafish, we described that MoS2 2D nanomaterials can reach, upon water (i.e. ambient) exposure, the nervous system of larvae, resulting in a direct neurological damage.
Collapse
Affiliation(s)
- Giuseppe Di Mauro
- Neuron Physiology and Technology Lab, Neuroscience area, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy.
| | - Viviana Jehová González
- Instituto Regional de Investigación Científica Aplicada (IRICA), UCLM, 13071 Ciudad Real, Spain
| | - Francesco Bambini
- Neuron Physiology and Technology Lab, Neuroscience area, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy.
| | - Silvia Camarda
- Neuron Physiology and Technology Lab, Neuroscience area, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy.
| | - Eduardo Prado
- Department of Applied Physics, Faculty of Science, University of Castilla La Mancha, Avda. Camilo José Cela 10, 13071 Ciudad Real, Spain
| | - Juan Pedro Holgado
- Instituto de Ciencia de Materiales de Sevilla, Centro Mixto Universidad de Sevilla-CSIC, Américo Vespucio, 49, 41092 Sevilla, Spain
| | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA), UCLM, 13071 Ciudad Real, Spain
- Facultad de Ciencias y Tecnologías Químicas, UCLM, Avda. Camilo José Cela S/N, Ciudad Real, Spain
| | - Laura Ballerini
- Neuron Physiology and Technology Lab, Neuroscience area, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy.
| | - Giada Cellot
- Neuron Physiology and Technology Lab, Neuroscience area, International School for Advanced Studies (SISSA), Via Bonomea 265, 34136, Trieste, Italy.
| |
Collapse
|
4
|
Firth W, Pye KR, Weightman Potter PG. Astrocytes at the intersection of ageing, obesity, and neurodegeneration. Clin Sci (Lond) 2024; 138:515-536. [PMID: 38652065 DOI: 10.1042/cs20230148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Once considered passive cells of the central nervous system (CNS), glia are now known to actively maintain the CNS parenchyma; in recent years, the evidence for glial functions in CNS physiology and pathophysiology has only grown. Astrocytes, a heterogeneous group of glial cells, play key roles in regulating the metabolic and inflammatory landscape of the CNS and have emerged as potential therapeutic targets for a variety of disorders. This review will outline astrocyte functions in the CNS in healthy ageing, obesity, and neurodegeneration, with a focus on the inflammatory responses and mitochondrial function, and will address therapeutic outlooks.
Collapse
Affiliation(s)
- Wyn Firth
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, U.K
| | - Katherine R Pye
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Paul G Weightman Potter
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| |
Collapse
|
5
|
Aktaş A, Yiğit F, Delibaş B, Kaplan AA, Hamour HM, Marangoz AH, Kaya A, Altun G, Kaplan S. The effects of Garcinia kola and curcumin on the dorsal root ganglion of the diabetic rat after peripheral nerve transection injury. J Chem Neuroanat 2024; 136:102395. [PMID: 38320670 DOI: 10.1016/j.jchemneu.2024.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/08/2024]
Abstract
OBJECTIVE To test the protective effects of Garcinia kola and curcumin on the ganglion tissues of diabetic rats following the use of autologous vein graft in peripheral nerve transection injury. METHODS The sciatic nerve on the right side was transected, and anastomosis was performed between the proximal and distal ends using an autologous vein graft. Curcumin and Garcinia kola seed extract were administered daily by oral gavage. The ganglion tissues were harvested after a 90-day waiting period. Sensory neurons in the dorsal root ganglion at the L4 and L5 levels were used for stereological evaluations. Mean sensory neuron numbers were analyzed using a stereological technique. The size of the light and dark neurons was also estimated, and ultrastructural and immunohistochemical evaluations were performed. RESULTS A statistically significant difference in sensory neuron numbers was observed between the groups with and without Garcinia kola and curcumin applications. The immunohistochemical results showed that the s-100 protein is expressed selectively between cell types. CONCLUSION The results of this study show that curcumin and Garicinia kola prevented sensory neuron loss in diabetic rats following transection injury to the sciatic nerve.
Collapse
Affiliation(s)
- Abit Aktaş
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University - Cerrahpaşa, Istanbul, Turkey
| | - Funda Yiğit
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University - Cerrahpaşa, Istanbul, Turkey
| | - Burcu Delibaş
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Arife Ahsen Kaplan
- Department of Histology and Embryology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Hala Mahgoub Hamour
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | | | - Ayşenur Kaya
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey; Department of Histology and Embryology, Faculty of Medicine, Karamanoğlu Mehmetbey University, Karaman, Turkey
| | - Gamze Altun
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Süleyman Kaplan
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey; Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania.
| |
Collapse
|
6
|
Brignone MS, Lanciotti A, Molinari P, Mallozzi C, De Nuccio C, Caprini ES, Petrucci TC, Visentin S, Ambrosini E. Megalencephalic leukoencephalopathy with subcortical cysts protein-1: A new calcium-sensitive protein functionally activated by endoplasmic reticulum calcium release and calmodulin binding in astrocytes. Neurobiol Dis 2024; 190:106388. [PMID: 38141856 DOI: 10.1016/j.nbd.2023.106388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/04/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND MLC1 is a membrane protein highly expressed in brain perivascular astrocytes and whose mutations account for the rare leukodystrophy (LD) megalencephalic leukoencephalopathy with subcortical cysts disease (MLC). MLC is characterized by macrocephaly, brain edema and cysts, myelin vacuolation and astrocyte swelling which cause cognitive and motor dysfunctions and epilepsy. In cultured astrocytes, lack of functional MLC1 disturbs cell volume regulation by affecting anion channel (VRAC) currents and the consequent regulatory volume decrease (RVD) occurring in response to osmotic changes. Moreover, MLC1 represses intracellular signaling molecules (EGFR, ERK1/2, NF-kB) inducing astrocyte activation and swelling following brain insults. Nevertheless, to date, MLC1 proper function and MLC molecular pathogenesis are still elusive. We recently reported that in astrocytes MLC1 phosphorylation by the Ca2+/Calmodulin-dependent kinase II (CaMKII) in response to intracellular Ca2+ release potentiates MLC1 activation of VRAC. These results highlighted the importance of Ca2+ signaling in the regulation of MLC1 functions, prompting us to further investigate the relationships between intracellular Ca2+ and MLC1 properties. METHODS We used U251 astrocytoma cells stably expressing wild-type (WT) or mutated MLC1, primary mouse astrocytes and mouse brain tissue, and applied biochemistry, molecular biology, video imaging and electrophysiology techniques. RESULTS We revealed that WT but not mutant MLC1 oligomerization and trafficking to the astrocyte plasma membrane is favored by Ca2+ release from endoplasmic reticulum (ER) but not by capacitive Ca2+ entry in response to ER depletion. We also clarified the molecular events underlining MLC1 response to cytoplasmic Ca2+ increase, demonstrating that, following Ca2+ release, MLC1 binds the Ca2+ effector protein calmodulin (CaM) at the carboxyl terminal where a CaM binding sequence was identified. Using a CaM inhibitor and generating U251 cells expressing MLC1 with CaM binding site mutations, we found that CaM regulates MLC1 assembly, trafficking and function, being RVD and MLC-linked signaling molecules abnormally regulated in these latter cells. CONCLUSION Overall, we qualified MLC1 as a Ca2+ sensitive protein involved in the control of volume changes in response to ER Ca2+ release and astrocyte activation. These findings provide new insights for the comprehension of the molecular mechanisms responsible for the myelin degeneration occurring in MLC and other LD where astrocytes have a primary role in the pathological process.
Collapse
Affiliation(s)
- M S Brignone
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - A Lanciotti
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - P Molinari
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - C Mallozzi
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - C De Nuccio
- Istituto Superiore di Sanità, Research Coordination and Support Service, Viale Regina Elena 299, 00161 Rome, Italy
| | - E S Caprini
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - T C Petrucci
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy
| | - S Visentin
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, Viale Regina Elena 299, 00161 Rome, Italy
| | - E Ambrosini
- Istituto Superiore di Sanità, Department of Neuroscience, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
7
|
Abd El-Rahman AA, El-Shafei SMA, Shehab GMG, Mansour L, Abuelsaad ASA, Gad RA. Assessment of Biochemical and Neuroactivities of Cultural Filtrate from Trichoderma harzianum in Adjusting Electrolytes and Neurotransmitters in Hippocampus of Epileptic Rats. Life (Basel) 2023; 13:1815. [PMID: 37763219 PMCID: PMC10533195 DOI: 10.3390/life13091815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Epilepsy is a serious chronic neurological disorder, which is accompanied by recurrent seizures. Repeated seizures cause physical injuries and neuronal dysfunction and may be a risk of cancer and vascular diseases. However, many antiepileptic drugs (AEDs) have side effects of mood alteration or neurocognitive function, a reduction in neuron excitation, and the inhibition of normal activity. Therefore, the present study aimed to evaluate the effect of secondary metabolites of Trichoderma harzianum cultural filtrate (ThCF) when adjusting different electrolytes and neurotransmitters in the hippocampus of epileptic rats. METHODS Cytotoxicity of ThCF against LS-174T cancer cells was assessed using a sulforhodamine B (SRB) assay. Quantitative estimation for some neurotransmitters, electrolytes in sera or homogenate of hippocampi tissues, and mRNA gene expression for ion or voltage gates was assessed by quantitative Real-Time PCR. RESULTS Treatment with ThCF reduces the proliferative percentage of LS-174T cells in a concentration-dependent manner. ThCF administration improves hyponatremia, hyperkalemia, and hypocalcemia in the sera of the epilepticus model. ThCF rebalances the elevated levels of many neurotransmitters and reduces the release of GABA and acetylcholine-esterase. Also, treatments with ThCF ameliorate the downregulation of mRNA gene expression for some gate receptors in hippocampal homogenate tissues and recorded a highly significant elevation in the expression of SCN1A, CACNA1S, and NMDA. CONCLUSION Secondary metabolites of Trichoderma (ThCF) have cytotoxic activity against LS-174T (colorectal cancer cell line) and anxiolytic-like activity through a GABAergic mechanism of action and an increase in GABA as inhibitory amino acid in the selected brain regions and reduced levels of NMDA and DOPA. The present data suggested that ThCF may inhibit intracellular calcium accumulation by triggering the NAADP-mediated Ca2+ signaling pathway. Therefore, the present results suggested further studies on the molecular pathway for each metabolite of ThCF, e.g., 6-pentyl-α-pyrone (6-PP), harzianic acid (HA), and hydrophobin, as an alternative drug to mitigate the side effects of AEDs.
Collapse
Affiliation(s)
- Atef A. Abd El-Rahman
- Department of Agricultural Chemistry, Faculty of Agriculture, Minia University, El-Minya 61519, Egypt;
| | - Sally M. A. El-Shafei
- Department of Agricultural Chemistry, Faculty of Agriculture, Minia University, El-Minya 61519, Egypt;
| | - Gaber M. G. Shehab
- Department of Biochemistry, Faculty of Agriculture, Cairo University, Giza 12613, Egypt;
| | - Lamjed Mansour
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Abdelaziz S. A. Abuelsaad
- Immunology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt;
| | - Rania A. Gad
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, NAHDA University (NUB), Beni-Suef 62511, Egypt;
| |
Collapse
|
8
|
Aten S, Du Y, Taylor O, Dye C, Collins K, Thomas M, Kiyoshi C, Zhou M. Chronic Stress Impairs the Structure and Function of Astrocyte Networks in an Animal Model of Depression. Neurochem Res 2023; 48:1191-1210. [PMID: 35796915 PMCID: PMC9823156 DOI: 10.1007/s11064-022-03663-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/18/2022] [Indexed: 01/11/2023]
Abstract
Now astrocytes appear to be the key contributors to the pathophysiology of major depression. Evidence in rodents shows that chronic stress is associated with a decreased expression of astrocytic GFAP-immunoreactivity within the cortex in addition to changes in the complexity and length of astrocyte processes. Furthermore, postmortem brains of individuals with depression have revealed a decrease in astrocyte density. Notably, astrocytes are extensively coupled to one another through gap junctions to form a network, or syncytium, and we have previously demonstrated that syncytial isopotentiality is a mechanism by which astrocytes function as an efficient system with respect to brain homeostasis. Interestingly, the question of how astrocyte network function changes following chronic stress is yet to be elucidated. Here, we sought to examine the effects of chronic stress on network-level astrocyte (dys)function. Using a transgenic aldh1l1-eGFP astrocyte reporter mouse, a six-week unpredictable chronic mild stress (UCMS) paradigm as a rodent model of major depression, and immunohistochemical approaches, we show that the morphology of individual astrocytes is altered by chronic stress exposure. Additionally, in astrocyte syncytial isopotentiality measurement, we found that UCMS impairs the syncytial coupling strength of astrocytes within the hippocampus and prefrontal cortex-two brain regions that have been implicated in the regulation of mood. Together, these findings reveal that chronic stress leads to astrocyte atrophy and impaired gap junction coupling, raising the prospect that both individual and network-level astrocyte functionality are important in the etiology of major depression and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sydney Aten
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
- Department of Neurology, Division of Sleep Medicine, and Program in Neuroscience, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yixing Du
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Olivia Taylor
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Courtney Dye
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Kelsey Collins
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Matthew Thomas
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
| | - Conrad Kiyoshi
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA
- Northern Marianas College, Saipan, MP, USA
| | - Min Zhou
- Department of Neuroscience, Ohio State University Wexner Medical Center, Graves Hall, Rm 4066C, 333 W. 10th Ave, Columbus, OH, 43210, USA.
| |
Collapse
|
9
|
Zhong S, Kiyoshi CM, Du Y, Wang W, Luo Y, Wu X, Taylor AT, Ma B, Aten S, Liu X, Zhou M. Genesis of a functional astrocyte syncytium in the developing mouse hippocampus. Glia 2023; 71:1081-1098. [PMID: 36598109 PMCID: PMC10777263 DOI: 10.1002/glia.24327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/03/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023]
Abstract
Astrocytes are increasingly shown to operate as an isopotential syncytium in brain function. Protoplasmic astrocytes acquire this ability to functionally go beyond the single-cell level by evolving into a spongiform morphology, cytoplasmically connecting into a syncytium, and expressing a high density of K+ conductance. However, none of these cellular/functional features exist in neonatal newborn astrocytes, which imposes a basic question of when a functional syncytium evolves in the developing brain. Our results show that the spongiform morphology of individual astrocytes and their spatial organization all reach stationary levels by postnatal day (P) 15 in the hippocampal CA1 region. Functionally, astrocytes begin to uniformly express a mature level of passive K+ conductance by P11. We next used syncytial isopotentiality measurement to monitor the maturation of the astrocyte syncytium. In uncoupled P1 astrocytes, the substitution of endogenous K+ by a Na+ -electrode solution ([Na+ ]p ) resulted in the total elimination of the physiological membrane potential (VM ), and outward K+ conductance as predicted by the Goldman-Hodgkin-Katz (GHK) equation. As more astrocytes are coupled to each other through gap junctions during development, the [Na+ ]p -induced loss of physiological VM and the outward K+ conductance is progressively compensated by the neighboring astrocytes. By P15, a stably established syncytial isopotentiality (-73 mV), and a fully compensated outward K+ conductance appeared in all [Na+ ]p -recorded astrocytes. Thus, in view of the developmental timeframe wherein a singular syncytium is anatomically and functionally established for intra-syncytium K+ equilibration, an astrocyte syncytium becomes fully operational at P15 in the mouse hippocampus.
Collapse
Affiliation(s)
- Shiying Zhong
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Neurology, Shanghai 10Hospital of Tongji University, School of Medicine, Shanghai, 200072, China
| | - Conrad M. Kiyoshi
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Yixing Du
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wei Wang
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Physiology, Tongji Medical College, Wuhan, 430030, China
| | - Yumeng Luo
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Xiao Wu
- Department of Neurology, Wuhan First Hospital, Wuhan 430022, China
| | - Anne T. Taylor
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Baofeng Ma
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sydney Aten
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Xueyuan Liu
- Department of Neurology, Shanghai 10Hospital of Tongji University, School of Medicine, Shanghai, 200072, China
| | - Min Zhou
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
10
|
Meyer E, Rieder P, Gobbo D, Candido G, Scheller A, de Oliveira RMW, Kirchhoff F. Cannabidiol Exerts a Neuroprotective and Glia-Balancing Effect in the Subacute Phase of Stroke. Int J Mol Sci 2022; 23:12886. [PMID: 36361675 PMCID: PMC9659180 DOI: 10.3390/ijms232112886] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 11/22/2022] Open
Abstract
Pharmacological agents limiting secondary tissue loss and improving functional outcomes after stroke are still limited. Cannabidiol (CBD), the major non-psychoactive component of Cannabis sativa, has been proposed as a neuroprotective agent against experimental cerebral ischemia. The effects of CBD mostly relate to the modulation of neuroinflammation, including glial activation. To investigate the effects of CBD on glial cells after focal ischemia in vivo, we performed time-lapse imaging of microglia and astroglial Ca2+ signaling in the somatosensory cortex in the subacute phase of stroke by in vivo two-photon laser-scanning microscopy using transgenic mice with microglial EGFP expression and astrocyte-specific expression of the genetically encoded Ca2+ sensor GCaMP3. CBD (10 mg/kg, intraperitoneally) prevented ischemia-induced neurological impairment, reducing the neurological deficit score from 2.0 ± 1.2 to 0.8 ± 0.8, and protected against neurodegeneration, as shown by the reduction (more than 70%) in Fluoro-Jade C staining (18.8 ± 7.5 to 5.3 ± 0.3). CBD reduced ischemia-induced microglial activation assessed by changes in soma area and total branch length, and exerted a balancing effect on astroglial Ca2+ signals. Our findings indicate that the neuroprotective effects of CBD may occur in the subacute phase of ischemia, and reinforce its strong anti-inflammatory property. Nevertheless, its mechanism of action on glial cells still requires further studies.
Collapse
Affiliation(s)
- Erika Meyer
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Phillip Rieder
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Gabriella Candido
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| | - Rúbia Maria Weffort de Oliveira
- Laboratory of Brain Ischemia and Neuroprotection, Department of Pharmacology and Therapeutics, State University of Maringá, Maringá 87020900, Brazil
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, D-66421 Homburg, Germany
| |
Collapse
|
11
|
Jehle A, Garaschuk O. The Interplay between cGMP and Calcium Signaling in Alzheimer's Disease. Int J Mol Sci 2022; 23:7048. [PMID: 35806059 PMCID: PMC9266933 DOI: 10.3390/ijms23137048] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic guanosine monophosphate (cGMP) is a ubiquitous second messenger and a key molecule in many important signaling cascades in the body and brain, including phototransduction, olfaction, vasodilation, and functional hyperemia. Additionally, cGMP is involved in long-term potentiation (LTP), a cellular correlate of learning and memory, and recent studies have identified the cGMP-increasing drug Sildenafil as a potential risk modifier in Alzheimer's disease (AD). AD development is accompanied by a net increase in the expression of nitric oxide (NO) synthases but a decreased activity of soluble guanylate cyclases, so the exact sign and extent of AD-mediated imbalance remain unclear. Moreover, human patients and mouse models of the disease present with entangled deregulation of both cGMP and Ca2+ signaling, e.g., causing changes in cGMP-mediated Ca2+ release from the intracellular stores as well as Ca2+-mediated cGMP production. Still, the mechanisms governing such interplay are poorly understood. Here, we review the recent data on mechanisms underlying the brain cGMP signaling and its interconnection with Ca2+ signaling. We also discuss the recent evidence stressing the importance of such interplay for normal brain function as well as in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
12
|
Rieder P, Gobbo D, Stopper G, Welle A, Damo E, Kirchhoff F, Scheller A. Astrocytes and Microglia Exhibit Cell-Specific Ca2+ Signaling Dynamics in the Murine Spinal Cord. Front Mol Neurosci 2022; 15:840948. [PMID: 35431801 PMCID: PMC9006623 DOI: 10.3389/fnmol.2022.840948] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 12/31/2022] Open
Abstract
The spinal cord is the main pathway connecting brain and peripheral nervous system. Its functionality relies on the orchestrated activity of both neurons and glial cells. To date, most advancement in understanding the spinal cord inner mechanisms has been made either by in vivo exposure of its dorsal surface through laminectomy or by acute ex vivo slice preparation, likely affecting spinal cord physiology in virtue of the necessary extensive manipulation of the spinal cord tissue. This is especially true of cells immediately responding to alterations of the surrounding environment, such as microglia and astrocytes, reacting within seconds or minutes and for up to several days after the original insult. Ca2+ signaling is considered one of the most immediate, versatile, and yet elusive cellular responses of glia. Here, we induced the cell-specific expression of the genetically encoded Ca2+ indicator GCaMP3 to evaluate spontaneous intracellular Ca2+ signaling in astrocytes and microglia. Ca2+ signals were then characterized in acute ex vivo (both gray and white matter) as well as in chronic in vivo (white matter) preparations using MSparkles, a MATLAB-based software for automatic detection and analysis of fluorescence events. As a result, we were able to segregate distinct astroglial and microglial Ca2+ signaling patterns along with method-specific Ca2+ signaling alterations, which must be taken into consideration in the reliable evaluation of any result obtained in physiological as well as pathological conditions. Our study revealed a high degree of Ca2+ signaling diversity in glial cells of the murine spinal cord, thus adding to the current knowledge of the astonishing glial heterogeneity and cell-specific Ca2+ dynamics in non-neuronal networks.
Collapse
Affiliation(s)
- Phillip Rieder
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Gebhard Stopper
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anna Welle
- Department of Genetics and Epigenetics, University of Saarland, Saarbrücken, Germany
| | - Elisa Damo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
- Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
- *Correspondence: Anja Scheller,
| |
Collapse
|
13
|
Orts-Del'Immagine A, Dhanasekar M, Lejeune FX, Roussel J, Wyart C. A norepinephrine-dependent glial calcium wave travels in the spinal cord upon acoustovestibular stimuli. Glia 2021; 70:491-507. [PMID: 34773299 DOI: 10.1002/glia.24118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023]
Abstract
Although calcium waves have been widely observed in glial cells, their occurrence in vivo during behavior remains less understood. Here, we investigated the recruitment of glial cells in the hindbrain and spinal cord after acousto-vestibular (AV) stimuli triggering escape responses using in vivo population calcium imaging in larval zebrafish. We observed that gap-junction-coupled spinal glial network exhibits large and homogenous calcium increases that rose in the rostral spinal cord and propagated bi-directionally toward the spinal cord and toward the hindbrain. Spinal glial calcium waves were driven by the recruitment of neurons and in particular, of noradrenergic signaling acting through α-adrenergic receptors. Noradrenergic neurons of the medulla-oblongata (NE-MO) were revealed in the vicinity of where the calcium wave started. NE-MO were recruited upon AV stimulation and sent dense axonal projections in the rostro-lateral spinal cord, suggesting these cells could trigger the glial wave to propagate down the spinal cord. Altogether, our results revealed that a simple AV stimulation is sufficient to recruit noradrenergic neurons in the brainstem that trigger in the rostral spinal cord two massive glial calcium waves, one traveling caudally in the spinal cord and another rostrally into the hindbrain.
Collapse
Affiliation(s)
| | | | | | | | - Claire Wyart
- Institut du cerveau, Sorbonne Université, Paris, France
| |
Collapse
|
14
|
McNeill J, Rudyk C, Hildebrand ME, Salmaso N. Ion Channels and Electrophysiological Properties of Astrocytes: Implications for Emergent Stimulation Technologies. Front Cell Neurosci 2021; 15:644126. [PMID: 34093129 PMCID: PMC8173131 DOI: 10.3389/fncel.2021.644126] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Astrocytes comprise a heterogeneous cell population characterized by distinct morphologies, protein expression and function. Unlike neurons, astrocytes do not generate action potentials, however, they are electrically dynamic cells with extensive electrophysiological heterogeneity and diversity. Astrocytes are hyperpolarized cells with low membrane resistance. They are heavily involved in the modulation of K+ and express an array of different voltage-dependent and voltage-independent channels to help with this ion regulation. In addition to these K+ channels, astrocytes also express several different types of Na+ channels; intracellular Na+ signaling in astrocytes has been linked to some of their functional properties. The physiological hallmark of astrocytes is their extensive intracellular Ca2+ signaling cascades, which vary at the regional, subregional, and cellular levels. In this review article, we highlight the physiological properties of astrocytes and the implications for their function and influence of network and synaptic activity. Furthermore, we discuss the implications of these differences in the context of optogenetic and DREADD experiments and consider whether these tools represent physiologically relevant techniques for the interrogation of astrocyte function.
Collapse
Affiliation(s)
- Jessica McNeill
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
15
|
Daliri EBM, Ofosu FK, Xiuqin C, Chelliah R, Oh DH. Probiotic Effector Compounds: Current Knowledge and Future Perspectives. Front Microbiol 2021; 12:655705. [PMID: 33746935 PMCID: PMC7965967 DOI: 10.3389/fmicb.2021.655705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/12/2021] [Indexed: 12/22/2022] Open
Abstract
Understanding the mechanism behind probiotic action will enable a rational selection of probiotics, increase the chances of success in clinical studies and make it easy to substantiate health claims. However, most probiotic studies over the years have rather focused on the effects of probiotics in health and disease, whereas little is known about the specific molecules that trigger effects in hosts. This makes it difficult to describe the detailed mechanism by which a given probiotic functions. Probiotics communicate with their hosts through molecular signaling. Meanwhile, since the molecules produced by probiotics under in vitro conditions may differ from those produced in vivo, in vitro mechanistic studies would have to be conducted under conditions that mimic gastrointestinal conditions as much as possible. The ideal situation would, however, be to carry out well-designed clinical trials in humans (or the target animal) using adequate quantities of the suspected probiotic molecule(s) or adequate quantities of isogenic knock-out or knock-in probiotic mutants. In this review, we discuss our current knowledge about probiotic bacteria and yeast molecules that are involved in molecular signaling with the host. We also discuss the challenges and future perspectives in the search for probiotic effector molecules.
Collapse
Affiliation(s)
- Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Fred Kwame Ofosu
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Chen Xiuqin
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
16
|
Signal transduction associated with lead-induced neurological disorders: A review. Food Chem Toxicol 2021; 150:112063. [PMID: 33596455 DOI: 10.1016/j.fct.2021.112063] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
Lead is a heavy metal pollutant that is widely present in the environment. It affects every organ system, yet the nervous system appears to be the most sensitive and primary target. Although many countries have made significant strides in controlling Pb pollution, Pb poisoning continuous to be a major public health concern. Exposure to Pb causes neurotoxicity that ranges from neurodevelopmental disorders to severe neurodegenerative lesions, leading to impairments in learning, memory, and cognitive function. Studies on the mechanisms of Pb-induced nervous system injury have convincingly shown that this metal can affect a plethora of cellular pathways affecting on cell survival, altering calcium dyshomeostasis, and inducing apoptosis, inflammation, energy metabolism disorders, oxidative stress, autophagy and glial stress. This review summarizes recent knowledge on multiple signaling pathways associated with Pb-induced neurological disorders in vivo and in vitro.
Collapse
|
17
|
Gamage R, Wagnon I, Rossetti I, Childs R, Niedermayer G, Chesworth R, Gyengesi E. Cholinergic Modulation of Glial Function During Aging and Chronic Neuroinflammation. Front Cell Neurosci 2020; 14:577912. [PMID: 33192323 PMCID: PMC7594524 DOI: 10.3389/fncel.2020.577912] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
Aging is a complex biological process that increases the risk of age-related cognitive degenerative diseases such as dementia, including Alzheimer’s disease (AD), Lewy Body Dementia (LBD), and mild cognitive impairment (MCI). Even non-pathological aging of the brain can involve chronic oxidative and inflammatory stress, which disrupts the communication and balance between the brain and the immune system. There has been an increasingly strong connection found between chronic neuroinflammation and impaired memory, especially in AD. While microglia and astrocytes, the resident immune cells of the central nervous system (CNS), exerting beneficial effects during the acute inflammatory phase, during chronic neuroinflammation they can become more detrimental. Central cholinergic circuits are involved in maintaining normal cognitive function and regulating signaling within the entire cerebral cortex. While neuronal-glial cholinergic signaling is anti-inflammatory and anti-oxidative, central cholinergic neuronal degeneration is implicated in impaired learning, memory sleep regulation, and attention. Although there is evidence of cholinergic involvement in memory, fewer studies have linked the cholinergic anti-inflammatory and anti-oxidant pathways to memory processes during development, normal aging, and disease states. This review will summarize the current knowledge of cholinergic effects on microglia and astroglia, and their role in both anti-inflammatory and anti-oxidant mechanisms, concerning normal aging and chronic neuroinflammation. We provided details on how stimulation of α7 nicotinic acetylcholine (α7nACh) receptors can be neuroprotective by increasing amyloid-β phagocytosis, decreasing inflammation and reducing oxidative stress by promoting the nuclear factor erythroid 2-related factor 2 (Nrf2) pathways and decreasing the release of pro-inflammatory cytokines. There is also evidence for astroglial α7nACh receptor stimulation mediating anti-inflammatory and antioxidant effects by inhibiting the nuclear factor-κB (NF-κB) pathway and activating the Nrf2 pathway respectively. We conclude that targeting cholinergic glial interactions between neurons and glial cells via α7nACh receptors could regulate neuroinflammation and oxidative stress, relevant to the treatment of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rashmi Gamage
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ingrid Wagnon
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ilaria Rossetti
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Ryan Childs
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Garry Niedermayer
- School of Science, Western Sydney University, Penrith, NSW, Australia
| | - Rose Chesworth
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
18
|
Rocchio F, Tapella L, Manfredi M, Chisari M, Ronco F, Ruffinatti FA, Conte E, Canonico PL, Sortino MA, Grilli M, Marengo E, Genazzani AA, Lim D. Gene expression, proteome and calcium signaling alterations in immortalized hippocampal astrocytes from an Alzheimer's disease mouse model. Cell Death Dis 2019; 10:24. [PMID: 30631041 PMCID: PMC6328590 DOI: 10.1038/s41419-018-1264-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 12/29/2022]
Abstract
Evidence is rapidly growing regarding a role of astroglial cells in the pathogenesis of Alzheimer’s disease (AD), and the hippocampus is one of the important brain regions affected in AD. While primary astroglial cultures, both from wild-type mice and from rodent models of AD, have been useful for studying astrocyte-specific alterations, the limited cell number and short primary culture lifetime have limited the use of primary hippocampal astrocytes. To overcome these limitations, we have now established immortalized astroglial cell lines from the hippocampus of 3xTg-AD and wild-type control mice (3Tg-iAstro and WT-iAstro, respectively). Both 3Tg-iAstro and WT-iAstro maintain an astroglial phenotype and markers (glutamine synthetase, aldehyde dehydrogenase 1 family member L1 and aquaporin-4) but display proliferative potential until at least passage 25. Furthermore, these cell lines maintain the potassium inward rectifying (Kir) current and present transcriptional and proteomic profiles compatible with primary astrocytes. Importantly, differences between the 3Tg-iAstro and WT-iAstro cell lines in terms of calcium signaling and in terms of transcriptional changes can be re-conducted to the changes previously reported in primary astroglial cells. To illustrate the versatility of this model we performed shotgun mass spectrometry proteomic analysis and found that proteins related to RNA binding and ribosome are differentially expressed in 3Tg-iAstro vs WT-iAstro. In summary, we present here immortalized hippocampal astrocytes from WT and 3xTg-AD mice that might be a useful model to speed up research on the role of astrocytes in AD.
Collapse
Affiliation(s)
- Francesca Rocchio
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy.,International Center for T1D, Pediatric Clinic Research Center Fondazione Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science L. Sacco, University of Milan, Milan, Italy
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Marcello Manfredi
- Department of Sciences and Technological Innovation, Università degli Studi del Piemonte Orientale, Alessandria, Italy.,ISALIT S.r.l., Spin-off of Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Mariangela Chisari
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia, 97, 95123, Catania, Italy
| | - Francesca Ronco
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy
| | | | - Eleonora Conte
- Department of Sciences and Technological Innovation, Università degli Studi del Piemonte Orientale, Alessandria, Italy
| | - Pier Luigi Canonico
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Maria Angela Sortino
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia, 97, 95123, Catania, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy
| | - Emilio Marengo
- Department of Sciences and Technological Innovation, Università degli Studi del Piemonte Orientale, Alessandria, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy.
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università degli Studi del Piemonte Orientale, Novara, Italy.
| |
Collapse
|
19
|
Clarke D, Penrose MA, Penstone T, Fuller-Carter PI, Hool LC, Harvey AR, Rodger J, Bates KA. Frequency-specific effects of repetitive magnetic stimulation on primary astrocyte cultures. Restor Neurol Neurosci 2018; 35:557-569. [PMID: 29172007 DOI: 10.3233/rnn-160708] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive technique that uses magnetic pulses over the cranium to induce electrical currents in underlying cortical tissue. Although rTMS has shown clinical utility for a number of neurological conditions, we have only limited understanding of how rTMS influences cellular function and cell-cell interactions. OBJECTIVE In this study, we sought to investigate whether repeated magnetic stimulation (rMS) can influence astrocyte biology in vitro. METHOD We tested four different rMS frequencies and measured the calcium response in primary neonatal astrocyte cultures. We also tested the effect of rMS on astrocyte migration and proliferation in vitro. We tested 3 to 4 culture replicates and 17 to 34 cells for each rMS frequency (sham, 1 Hz, cTBS, 10 Hz and biomemetic high frequency stimulation - BHFS). RESULTS Of all frequencies tested, 1 Hz stimulation resulted in a statistically significant rise in intracellular calcium in the cytoplasmic and nuclear compartments of the cultured astrocytes. This calcium rise did not affect migration or proliferation in the scratch assay, though astrocyte hypertrophy was reduced in response to 1 Hz rMS, 24 hours post scratch injury. CONCLUSION Our results provide preliminary evidence that rMS can influence astrocyte physiology, indicating the potential for a novel mechanism by which rTMS can influence brain activity.
Collapse
Affiliation(s)
- Darren Clarke
- School of Biological Sciences, The University of Western Australia, Perth, Crawley, WA, Australia.,School of Human Sciences, The University of Western Australia, Perth, Crawley, WA, Australia
| | - Marissa A Penrose
- School of Biological Sciences, The University of Western Australia, Perth, Crawley, WA, Australia
| | - Tamasin Penstone
- School of Biological Sciences, The University of Western Australia, Perth, Crawley, WA, Australia
| | - Paula I Fuller-Carter
- School of Biological Sciences, The University of Western Australia, Perth, Crawley, WA, Australia
| | - Livia C Hool
- School of Human Sciences, The University of Western Australia, Perth, Crawley, WA, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, Perth, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Perth, Crawley, WA, Australia.,Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Kristyn A Bates
- School of Biological Sciences, The University of Western Australia, Perth, Crawley, WA, Australia
| |
Collapse
|
20
|
Abstract
Spinal muscular atrophy (SMA) is a motoneuron disease caused by loss or mutation in Survival of Motor Neuron 1 (SMN1) gene. Recent studies have shown that selective restoration of SMN protein in astrocytes partially alleviates pathology in an SMA mouse model, suggesting important roles for astrocytes in SMA. Addressing these underlying mechanisms may provide new therapeutic avenues to fight SMA. Using primary cultures of pure motoneurons or astrocytes from SMNΔ7 (SMA) and wild-type (WT) mice, as well as their mixed and matched cocultures, we characterized the contributions of motoneurons, astrocytes, and their interactions to synapse loss in SMA. In pure motoneuron cultures, SMA motoneurons exhibited normal survival but intrinsic defects in synapse formation and synaptic transmission. In pure astrocyte cultures, SMA astrocytes exhibited defects in calcium homeostasis. In motoneuron-astrocyte contact cocultures, synapse formation and synaptic transmission were significantly reduced when either motoneurons, astrocytes or both were from SMA mice compared with those in WT motoneurons cocultured with WT astrocytes. The reduced synaptic activity is unlikely due to changes in motoneuron excitability. This disruption in synapse formation and synaptic transmission by SMN deficiency was not detected in motoneuron-astrocyte noncontact cocultures. Additionally, we observed a downregulation of Ephrin B2 in SMA astrocytes. These findings suggest that there are both cell autonomous and non-cell-autonomous defects in SMA motoneurons and astrocytes. Defects in contact interactions between SMA motoneurons and astrocytes impair synaptogenesis seen in SMA pathology, possibly due to the disruption of the Ephrin B2 pathway.
Collapse
|
21
|
Petit JM, Magistretti P. Regulation of neuron–astrocyte metabolic coupling across the sleep–wake cycle. Neuroscience 2016; 323:135-56. [DOI: 10.1016/j.neuroscience.2015.12.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 11/30/2022]
|
22
|
Role of transient receptor potential ankyrin 1 channels in Alzheimer's disease. J Neuroinflammation 2016; 13:92. [PMID: 27121378 PMCID: PMC4847235 DOI: 10.1186/s12974-016-0557-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 04/19/2016] [Indexed: 12/14/2022] Open
Abstract
Background Transient receptor potential ankyrin 1 (TRPA1) channel plays an important role in pain and inflammation. However, little is known about the significance of the TRPA1 channel in the pathophysiology of Alzheimer’s disease (AD). Methods Wild-type (WT), TRPA1−/−, amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic (APP/PS1 Tg) mice, the mouse model of AD, and APP/PS1 Tg/TRPA1−/− mice were used to examine the role of TRPA1 in pathogenesis of AD. Western blot was used for protein expression; immunohistochemistry was used for histological examination. The mouse behaviors were evaluated by locomotion, nesting building, Y-maze and Morris water maze tests; levels of interleukin (IL)-1β, IL-4, IL-6 and IL-10 and the activities of protein phosphatase 2B (PP2B), NF-κB and nuclear factor of activated T cells (NFAT) were measured by conventional assay kits; Fluo-8 NW calcium (Ca2+) assay kit was used for the measurement of intracellular Ca2+ level in primary astrocytes and HEK293 cells. Results The protein expression of TRPA1 channels was higher in brains, mainly astrocytes of the hippocampus, from APP/PS1 Tg mice than WT mice. Ablation of TRPA1-channel function in APP/PS1 Tg mice alleviated behavioral dysfunction, Aβ plaque deposition and pro-inflammatory cytokine production but increased astrogliosis in brain lesions. TRPA1 channels were activated and Ca2+ influx was elicited in both astrocytes and TRPA1-transfected HEK293 cells treated with fibrilized Aβ1–42; these were abrogated by pharmacological inhibition of TRPA1 channel activity, disruption of TRPA1 channel function or removal of extracellular Ca2+. Inhibition of TRPA1 channel activity exacerbated Aβ1–42–induced astrogliosis but inhibited Aβ1–42–increased PP2B activation, the production of pro-inflammatory cytokines and activities of transcriptional factors NF-κB and NFAT in astrocytes and in APP/PS1 Tg mice. Pharmacological inhibition of PP2B activity diminished the fibrilized Aβ1–42–induced production of pro-inflammatory cytokines, activation of NF-κB and NFAT and astrogliosis in astrocytes. Conclusions TRPA1 − Ca2+ − PP2B signaling may play a crucial role in regulating astrocyte-derived inflammation and pathogenesis of AD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0557-z) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Gao K, Wang CR, Jiang F, Wong AYK, Su N, Jiang JH, Chai RC, Vatcher G, Teng J, Chen J, Jiang YW, Yu ACH. Traumatic scratch injury in astrocytes triggers calcium influx to activate the JNK/c-Jun/AP-1 pathway and switch on GFAP expression. Glia 2013; 61:2063-77. [PMID: 24123203 DOI: 10.1002/glia.22577] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/11/2013] [Accepted: 08/21/2013] [Indexed: 01/25/2023]
Abstract
Astrocyte activation is a hallmark of central nervous system injuries resulting in glial scar formation (astrogliosis). The activation of astrocytes involves metabolic and morphological changes with complex underlying mechanisms, which should be defined to provide targets for astrogliosis intervention. Astrogliosis is usually accompanied by an upregulation of glial fibrillary acidic protein (GFAP). Using an in vitro scratch injury model, we scratched primary cultures of cerebral cortical astrocytes and observed an influx of calcium in the form of waves spreading away from the wound through gap junctions. Using the calcium blocker BAPTA-AM and the JNK inhibitor SP600125, we demonstrated that the calcium wave triggered the activation of JNK, which then phosphorylated the transcription factor c-Jun to facilitate the binding of AP-1 to the GFAP gene promoter to switch on GFAP upregulation. Blocking calcium mobilization with BAPTA-AM in an in vivo stab wound model reduced GFAP expression and glial scar formation, showing that the calcium signal, and the subsequent regulation of downstream signaling molecules, plays an essential role in brain injury response. Our findings demonstrated that traumatic scratch injury to astrocytes triggered a calcium influx from the extracellular compartment and activated the JNK/c-Jun/AP-1 pathway to switch on GFAP expression, identifying a previously unreported signaling cascade that is important in astrogliosis and the physiological response following brain injury.
Collapse
Affiliation(s)
- Kai Gao
- Neuroscience Research Institute, Key Laboratory for Neuroscience (Ministry of Education), Key Laboratory for Neuroscience (National Health and Family Planning Commission), Department of Neurobiology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Lunardi P, Nardin P, Guerra MC, Abib R, Leite MC, Gonçalves CA. Huperzine A, but not tacrine, stimulates S100B secretion in astrocyte cultures. Life Sci 2013; 92:701-7. [DOI: 10.1016/j.lfs.2013.01.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/11/2013] [Accepted: 01/25/2013] [Indexed: 01/09/2023]
|
25
|
Rozanski GM, Li Q, Kim H, Stanley EF. Purinergic transmission and transglial signaling between neuron somata in the dorsal root ganglion. Eur J Neurosci 2012; 37:359-65. [PMID: 23216714 DOI: 10.1111/ejn.12082] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 10/23/2012] [Accepted: 11/06/2012] [Indexed: 01/07/2023]
Abstract
Most dorsal root ganglion neuronal somata (NS) are isolated from their neighbours by a satellite glial cell (SGC) sheath. However, some NS are associated in pairs, separated solely by the membrane septum of a common SGC to form a neuron-glial cell-neuron (NGlN) trimer. We reported that stimulation of one NS evokes a delayed, noisy and long-duration inward current in both itself and its passive partner that was blocked by suramin, a general purinergic antagonist. Here we test the hypothesis that NGlN transmission involves purinergic activation of the SGC. Stimulation of the NS triggered a sustained current noise in the SGC. Block of transmission through the NGlN by reactive blue 2 or thapsigargin, a Ca(2+) store-depletion agent, implicated a Ca(2+) store discharge-linked P2Y receptor. P2Y2 was identified by simulation of the NGlN-like transmission by puffing UTP onto the SGC and by immunocytochemical localization to the SGC membrane septum. Block of the UTP effect by BAPTA, an intracellular Ca(2+) scavenger, supported the involvement of SGC Ca(2+) stores in the signaling pathway. We infer that transmission through the NGlN trimer involves secretion of ATP from the NS and triggering of SGC Ca(2+) store discharge via P2Y2 receptors. Presumably, cytoplasmic Ca(2+) elevation leads to the release of an as-yet unidentified second transmitter from the glial cell to complete transmission. Thus, the two NS of the NGlN trimer communicate via a 'sandwich synapse' transglial pathway, a novel signaling mechanism that may contribute to information transfer in other regions of the nervous system.
Collapse
Affiliation(s)
- Gabriela M Rozanski
- Laboratory of Synaptic Transmission, Toronto Western Research Institute, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
26
|
Molnár T, Héja L, Emri Z, Simon A, Nyitrai G, Pál I, Kardos J. Activation of astroglial calcium signaling by endogenous metabolites succinate and gamma-hydroxybutyrate in the nucleus accumbens. FRONTIERS IN NEUROENERGETICS 2011; 3:7. [PMID: 22180742 PMCID: PMC3235779 DOI: 10.3389/fnene.2011.00007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 11/25/2011] [Indexed: 11/18/2022]
Abstract
Accumulating evidence suggests that different energy metabolites play a role not only in neuronal but also in glial signaling. Recently, astroglial Ca2+ transients evoked by the major citric acid cycle metabolite succinate (SUC) and gamma-hydroxybutyrate (GHB) that enters the citric acid cycle via SUC have been described in the brain reward area, the nucleus accumbens (NAc). Cells responding to SUC by Ca2+ transient constitute a subset of ATP-responsive astrocytes that are activated in a neuron-independent way. In this study we show that GHB-evoked Ca2+ transients were also found to constitute a subset of ATP-responsive astrocytes in the NAc. Repetitive Ca2+ dynamics evoked by GHB suggested that Ca2+ was released from internal stores. Similarly to SUC, the GHB response was also characterized by an effective concentration of 50 μM. We observed that the number of ATP-responsive cells decreased with increasing concentration of either SUC or GHB. Moreover, the concentration dependence of the number of ATP-responsive cells were highly identical as a function of both [SUC] and [GHB], suggesting a mutual receptor for SUC and GHB, therefore implying the existence of a distinct GHB-recognizing astroglial SUC receptor in the brain. The SUC-evoked Ca2+ signal remained in mice lacking GABAB receptor type 1 subunit in the presence and absence of the N-Methyl-d-Aspartate (NMDA) receptor antagonist (2R)-amino-5-phosphonovaleric acid (APV), indicating action mechanisms independent of the GABAB or NMDA receptor subtypes. By molecular docking calculations we found that residues R99, H103, R252, and R281 of the binding crevice of the kidney SUC-responsive membrane receptor SUCNR1 (GPCR91) also predict interaction with GHB, further implying similar GHB and SUC action mechanisms. We conclude that the astroglial action of SUC and GHB may represent a link between brain energy states and Ca2+ signaling in astrocytic networks.
Collapse
Affiliation(s)
- Tünde Molnár
- Department of Neurochemistry, Chemical Research Center, Hungarian Academy of Sciences Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
27
|
Molnár T, Dobolyi A, Nyitrai G, Barabás P, Héja L, Emri Z, Palkovits M, Kardos J. Calcium signals in the nucleus accumbens: activation of astrocytes by ATP and succinate. BMC Neurosci 2011; 12:96. [PMID: 21967230 PMCID: PMC3199278 DOI: 10.1186/1471-2202-12-96] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 10/03/2011] [Indexed: 12/13/2022] Open
Abstract
Background Accumulating evidence suggests that glial signalling is activated by different brain functions. However, knowledge regarding molecular mechanisms of activation or their relation to neuronal activity is limited. The purpose of the present study is to identify the characteristics of ATP-evoked glial signalling in the brain reward area, the nucleus accumbens (NAc), and thereby to explore the action of citric acid cycle intermediate succinate (SUC). Results We described the burst-like propagation of Ca2+ transients evoked by ATP in acute NAc slices from rat brain. Co-localization of the ATP-evoked Ca2+ signalling with immunoreactivities of the astroglia-specific gap junction forming channel protein connexin43 (Cx43) and the glial fibrillary acidic protein (GFAP) indicated that the responsive cells were a subpopulation of Cx43 and GFAP immunoreactive astrocytes. The ATP-evoked Ca2+ transients were present under the blockade of neuronal activity, but were inhibited by Ca2+ store depletion and antagonism of the G protein coupled purinergic P2Y1 receptor subtype-specific antagonist MRS2179. Similarly, Ca2+ transients evoked by the P2Y1 receptor subtype-specific agonist 2-(Methylthio)adenosine 5'-diphosphate were also blocked by MRS2179. These characteristics implied that intercellular Ca2+ signalling originated from the release of Ca2+ from internal stores, triggered by the activation of P2Y1 receptors. Inhibition by the gap junction blockers carbenoxolone and flufenamic acid and by an antibody raised against the gating-associated segment of Cx43 suggested that intercellular Ca2+ signalling proceeded through gap junctions. We demonstrated for the first time that extracellular SUC also evoked Ca2+ transients (EC50 = 50-60 μM) in about 15% of the ATP-responsive NAc astrocytes. By contrast to glial cells, electrophysiologically identified NAc neurons surrounded by ATP-responsive astrocytes were not activated simultaneously. Conclusions We concluded, therefore, that ATP- and SUC-sensitive Ca2+ transients appear to represent a signalling layer independent of NAc neurons. This previously unrecognised glial action of SUC, a major cellular energy metabolite, may play a role in linking metabolism to Ca2+ signalling in astrocytic networks under physiological and pathological conditions such as exercise and metabolic diseases.
Collapse
Affiliation(s)
- Tünde Molnár
- Department of Neurochemistry, Institute of Biomolecular Chemistry, Chemical Research Center, Hungarian Academy of Sciences, Pusztaszeriút 59-67, 1025 Budapest, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Microglial cells are the resident macrophages in the central nervous system. These cells of mesodermal/mesenchymal origin migrate into all regions of the central nervous system, disseminate through the brain parenchyma, and acquire a specific ramified morphological phenotype termed "resting microglia." Recent studies indicate that even in the normal brain, microglia have highly motile processes by which they scan their territorial domains. By a large number of signaling pathways they can communicate with macroglial cells and neurons and with cells of the immune system. Likewise, microglial cells express receptors classically described for brain-specific communication such as neurotransmitter receptors and those first discovered as immune cell-specific such as for cytokines. Microglial cells are considered the most susceptible sensors of brain pathology. Upon any detection of signs for brain lesions or nervous system dysfunction, microglial cells undergo a complex, multistage activation process that converts them into the "activated microglial cell." This cell form has the capacity to release a large number of substances that can act detrimental or beneficial for the surrounding cells. Activated microglial cells can migrate to the site of injury, proliferate, and phagocytose cells and cellular compartments.
Collapse
|
29
|
Van den Eynden J, Notelaers K, Brône B, Janssen D, Nelissen K, Sahebali S, Smolders I, Hellings N, Steels P, Rigo JM. Glycine enhances microglial intracellular calcium signaling. A role for sodium-coupled neutral amino acid transporters. Pflugers Arch 2011; 461:481-91. [PMID: 21350800 DOI: 10.1007/s00424-011-0939-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 01/31/2011] [Accepted: 02/09/2011] [Indexed: 12/17/2022]
Abstract
The inhibitory neurotransmitter glycine is known to enhance microglial nitric oxide production. However, up to now, the mechanism is undocumented. Since calcium is an important second messenger in both immune and glial cells, we studied the effects of glycine on intracellular calcium signaling. We found that millimolar concentrations of glycine enhance microglial intracellular calcium transients induced by 100 μM ATP or by 500 nM thapsigargin. This modulation was unaffected by the glycine receptor antagonist strychnine and could not be mimicked by glycine receptor agonists such as taurine or β-alanine, indicating glycine receptor independency. The modulation of calcium responses could be mimicked by several structurally related amino acids (e.g., serine, alanine, or glutamine) and was inhibited in the presence of the neutral amino acid transporter substrate α-aminoisobutyric acid (AIB). We correlated these findings to immunofluorescence glycine uptake experiments which showed a clear glycine uptake which was inhibited by AIB. Furthermore, all amino acids that were shown to modulate calcium responses also evoked AIB-sensitive inward currents, mainly carried by sodium, as demonstrated by patch clamp experiments. Based on these findings, we propose that sodium-coupled neutral amino acid transporters are responsible for the observed glycine modulation of intracellular calcium responses.
Collapse
Affiliation(s)
- Jimmy Van den Eynden
- Institute of Biomedical Research, Hasselt University and Transnationale Universiteit Limburg, Agoralaan, Diepenbeek, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Samuels SE, Lipitz JB, Dahl G, Muller KJ. Neuroglial ATP release through innexin channels controls microglial cell movement to a nerve injury. ACTA ACUST UNITED AC 2011; 136:425-42. [PMID: 20876360 PMCID: PMC2947054 DOI: 10.1085/jgp.201010476] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Microglia, the immune cells of the central nervous system, are attracted to sites of injury. The injury releases adenosine triphosphate (ATP) into the extracellular space, activating the microglia, but the full mechanism of release is not known. In glial cells, a family of physiologically regulated unpaired gap junction channels called innexons (invertebrates) or pannexons (vertebrates) located in the cell membrane is permeable to ATP. Innexons, but not pannexons, also pair to make gap junctions. Glial calcium waves, triggered by injury or mechanical stimulation, open pannexon/innexon channels and cause the release of ATP. It has been hypothesized that a glial calcium wave that triggers the release of ATP causes rapid microglial migration to distant lesions. In the present study in the leech, in which a single giant glial cell ensheathes each connective, hydrolysis of ATP with 10 U/ml apyrase or block of innexons with 10 µM carbenoxolone (CBX), which decreased injury-induced ATP release, reduced both movement of microglia and their accumulation at lesions. Directed movement and accumulation were restored in CBX by adding ATP, consistent with separate actions of ATP and nitric oxide, which is required for directed movement but does not activate glia. Injection of glia with innexin2 (Hminx2) RNAi inhibited release of carboxyfluorescein dye and microglial migration, whereas injection of innexin1 (Hminx1) RNAi did not when measured 2 days after injection, indicating that glial cells’ ATP release through innexons was required for microglial migration after nerve injury. Focal stimulation either mechanically or with ATP generated a calcium wave in the glial cell; injury caused a large, persistent intracellular calcium response. Neither the calcium wave nor the persistent response required ATP or its release. Thus, in the leech, innexin membrane channels releasing ATP from glia are required for migration and accumulation of microglia after nerve injury.
Collapse
Affiliation(s)
- Stuart E Samuels
- Neuroscience Program, University of Miami School of Medicine, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
31
|
Buibas M, Silva GA. A framework for simulating and estimating the state and functional topology of complex dynamic geometric networks. Neural Comput 2010; 23:183-214. [PMID: 20964542 DOI: 10.1162/neco_a_00065] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
We introduce a framework for simulating signal propagation in geometric networks (networks that can be mapped to geometric graphs in some space) and developing algorithms that estimate (i.e., map) the state and functional topology of complex dynamic geometric networks. Within the framework, we define the key features typically present in such networks and of particular relevance to biological cellular neural networks: dynamics, signaling, observation, and control. The framework is particularly well suited for estimating functional connectivity in cellular neural networks from experimentally observable data and has been implemented using graphics processing unit high-performance computing. Computationally, the framework can simulate cellular network signaling close to or faster than real time. We further propose a standard test set of networks to measure performance and compare different mapping algorithms.
Collapse
|
32
|
Glia: the many ways to modulate synaptic plasticity. Neurochem Int 2010; 57:440-5. [PMID: 20193723 DOI: 10.1016/j.neuint.2010.02.013] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 02/15/2010] [Accepted: 02/19/2010] [Indexed: 11/21/2022]
Abstract
Synaptic plasticity consists in a change in synaptic strength that is believed to be the basis of learning and memory. Synaptic plasticity has been for a very long period of time a hallmark of neurons. Recent advances in physiology of glial cells indicate that astrocyte and microglia possess all the features to participate and modulate the various form of synaptic plasticity. Indeed beside their respective supportive and immune functions an increasing number of study demonstrate that astrocytes and microglia express receptors for most neurotransmitters and release neuroactive substances that have been shown to modulate neuronal activity and synaptic plasticity. Because glial cells are all around synapses and release a wide variety of neuroactive molecule during physiological and pathological conditions, glial cells have been reported to modulate synaptic plasticity in many different ways. From change in synaptic coverage, to release of chemokines and cytokines up to dedicated "glio" transmitters release, glia were reported to affect synaptic scaling, homeostatic plasticity, metaplasticity, long-term potentiation and long-term depression.
Collapse
|
33
|
New concepts regarding cerebral vasospasm: glial-centric mechanisms. Can J Anaesth 2010; 57:479-89. [PMID: 20131107 DOI: 10.1007/s12630-010-9271-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Accepted: 01/12/2010] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Poor outcome in patients with cerebral vasospasm following subarachnoid hemorrhage remains a serious clinical problem. The current management with focus on the cerebrovascular constriction accounts for the use of "triple-H" therapy (hypertension, hypervolemia, and hemodilution) to enhance cerebral blood flow through constricted vessels. Recent work suggests that spreading depression (a stereotypical response of cerebral cortical tissue to noxious stimuli with subsequent oligemic blood flow) occurs in patients with cerebral vasospasm. A narrative review was conducted to examine the relationship between spreading depression and subarachnoid hemorrhage and to identify the anesthetic effects on the propagation of spreading depression. PRINCIPAL FINDINGS Following review of the literature, an underlying mechanism is advanced that cerebral vasospasm is not primarily a problem of the cerebral vasculature but a consequence of glial cell dysfunction following spreading depression - a glial-centric cause for vasospasm. Such a mechanism for vasospasm becomes manifest when spreading depression waves transition to peri-infarct depolarization waves - with protracted ischemic blood flow in compromised tissue. The extracellular microenvironment with high potassium and low nitric oxide tension can account for conducting vessel narrowing. CONCLUSIONS The implication for clinical management is discussed supposing glial cell dysfunction is an underlying mechanism responsible for the vascular spasm.
Collapse
|
34
|
Calcium homeostasis of acutely denervated and lesioned dentate gyrus in organotypic entorhino-hippocampal co-cultures. Cell Calcium 2010; 47:242-52. [PMID: 20053446 DOI: 10.1016/j.ceca.2009.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 11/04/2009] [Accepted: 12/07/2009] [Indexed: 12/20/2022]
Abstract
Denervation of neurons, e.g. upon traumatic injury or neuronal degeneration, induces transneuronal degenerative events, such as spine loss, dendritic pruning, and even cell loss. We studied one possible mechanism proposed to trigger such events, i.e. excess glutamate release from severed axons conveyed transsynaptically via postsynaptic calcium influx. Using 2-photon microscopical calcium imaging in organotypic entorhino-hippocampal co-cultures, we show that acute transection of the perforant path elicits two independent effects on calcium homeostasis in the dentate gyrus: a brief, short-latency elevation of postsynaptic calcium levels in denervated granule cells, which can be blocked by preincubation with tetrodotoxin, and a long-latency astroglial calcium wave, not blocked by tetrodotoxin and propagating slowly through the hippocampus. While neuronal calcium elevations upon axonal transection placed remote from the target area were similar to those elicited by brief trains of electrical stimulation of the perforant path, large-scale calcium signals were observed upon lesions placed close to or within the dendritic field of granule cells. Concordantly, induction of c-fos in denervated neurons coincided spatially with cell populations showing prolonged calcium elevations upon concomitant dendritic damage. Since denervation of dentate granule cells by remote transection of the perforant path induces transsynaptic dendritic reorganization in the utilized organotypic cultures, a generalized breakdown of the cellular calcium homeostasis is unlikely to underlie these transneuronal changes.
Collapse
|
35
|
Abstract
Astrocytes possess GPCRs (G-protein-coupled receptors) for neuroactive substances and can respond via these receptors to signals originating from neurons as well as astrocytes. Like many transmembrane proteins, GPCRs exist in a dynamic equilibrium between receptors expressed at the plasma membrane and those present within intracellular trafficking compartments. The characteristics of GPCR trafficking within astrocytes have not been investigated. We therefore monitored the trafficking of recombinant fluorescent protein chimeras of the CB1R (cannabinoid receptor 1) that is thought to be expressed natively in astrocytes. CB1R chimeras displayed a marked punctate intracellular localization when expressed in cultured rat visual cortex astrocytes, an expression pattern reminiscent of native CB1R expression in these cells. Based upon trafficking characteristics, we found the existence of two populations of vesicular CB1R puncta: (i) relatively immobile puncta with movement characteristic of diffusion and (ii) mobile puncta with movement characteristic of active transport along cytoskeletal elements. The predominant direction of active transport is oriented radially to/from the nuclear region, which can be abolished by disruption of the microtubule cytoskeleton. CB1R puncta are localized within intracellular acidic organelles, mainly co-localizing with endocytic compartments. Constitutive trafficking of CB1R to and from the plasma membrane is an energetically costly endeavour whose function is at present unclear in astrocytes. However, given that intracellular CB1Rs can engage cell signalling pathways, it is likely that this process plays an important regulatory role.
Collapse
|
36
|
The trinity of Ca2+ sources for the exocytotic glutamate release from astrocytes. Neurochem Int 2009; 55:2-8. [PMID: 19171170 DOI: 10.1016/j.neuint.2008.12.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2008] [Revised: 12/18/2008] [Accepted: 12/19/2008] [Indexed: 01/23/2023]
Abstract
Astrocytes can exocytotically release the transmitter glutamate. Increased cytosolic Ca(2+) concentration is necessary and sufficient in this process. The source of Ca(2+) for the Ca(2+)-dependent exocytotic release of glutamate from astrocytes predominately comes from endoplasmic reticulum (ER) stores with contributions from both inositol 1,4,5-trisphosphate- and ryanodine/caffeine-sensitive stores. An additional source of Ca(2+) comes from the extracellular space via store-operated Ca(2+) entry due to the depletion of ER stores. Here transient receptor potential canonical type 1 containing channels permit entry of Ca(2+) to the cytosol, which can then be transported by the store-specific Ca(2+)-ATPase to (re)fill ER. Mitochondria can modulate cytosolic Ca(2+) levels by affecting two aspects of the cytosolic Ca(2+) kinetics in astrocytes. They play a role in immediate sequestration of Ca(2+) during the cytosolic Ca(2+) increase in stimulated astrocytes as a result of Ca(2+) entry into the cytosol from ER stores and/or extracellular space. As cytosolic Ca(2+)declines due to activity of pumps, such as the smooth ER Ca(2+)-ATPase, free Ca(2+) is slowly released by mitochondria into cytosol. Taken together, the trinity of Ca(2+) sources, ER, extracellular space and mitochondria, can vary concentration of cytosolic Ca(2+) which in turn can modulate Ca(2+)-dependent vesicular glutamate release from astrocytes. An understanding of how these Ca(2+) sources contribute to glutamate release in (patho)physiology of astrocytes will provide information on astrocytic functions in health and disease and may also open opportunities for medical intervention.
Collapse
|
37
|
The role of the t-SNARE SNAP-25 in action potential-dependent calcium signaling and expression in GABAergic and glutamatergic neurons. BMC Neurosci 2008; 9:105. [PMID: 18959796 PMCID: PMC2600647 DOI: 10.1186/1471-2202-9-105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Accepted: 10/29/2008] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, comprised of SNAP-25, syntaxin 1A, and VAMP-2, has been shown to be responsible for action potential (AP)-dependent, calcium-triggered release of several neurotransmitters. However, this basic fusogenic protein complex may be further specialized to suit the requirements for different neurotransmitter systems, as exemplified by neurons and neuroendocrine cells. In this study, we investigate the effects of SNAP-25 ablation on spontaneous neuronal activity and the expression of functionally distinct isoforms of this t-SNARE in GABAergic and glutamatergic neurons of the adult brain. RESULTS We found that neurons cultured from Snap25 homozygous null mutant (Snap25-/-) mice failed to develop synchronous network activity seen as spontaneous AP-dependent calcium oscillations and were unable to trigger glial transients following depolarization. Voltage-gated calcium channel (VGCC) mediated calcium transients evoked by depolarization, nevertheless, did not differ between soma of SNAP-25 deficient and control neurons. Furthermore, we observed that although the expression of SNAP-25 RNA transcripts varied among neuronal populations in adult brain, the relative ratio of the transcripts encoding alternatively spliced SNAP-25 variant isoforms was not different in GABAergic and glutamatergic neurons. CONCLUSION We propose that the SNAP-25b isoform is predominantly expressed by both mature glutamatergic and GABAergic neurons and serves as a fundamental component of SNARE complex used for fast synaptic communication in excitatory and inhibitory circuits required for brain function. Moreover, SNAP-25 is required for neurons to establish AP-evoked synchronous network activity, as measured by calcium transients, whereas the loss of this t-SNARE does not affect voltage-dependent calcium entry.
Collapse
|
38
|
Pryazhnikov E, Khiroug L. Sub-micromolar increase in [Ca(2+)](i) triggers delayed exocytosis of ATP in cultured astrocytes. Glia 2008; 56:38-49. [PMID: 17910050 DOI: 10.1002/glia.20590] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Astrocytes release a variety of transmitter molecules, which mediate communication between glial cells in the brain and modulate synaptic transmission. ATP is a major glia-derived transmitter, but the mechanisms and kinetics of ATP release from astrocytes remain largely unknown. Here, we combined epifluorescence and total internal reflection fluorescence microscopy to monitor individual quinacrine-loaded ATP-containing vesicles undergoing exocytosis in cultured astrocytes. In resting cells, vesicles exhibited three-dimensional motility, spontaneous docking and release at low rate. Extracellular ATP application induced a Ca(2+)-dependent increase in the rate of exocytosis, which persisted for several minutes. Using UV flash photolysis of caged Ca(2+), the threshold [Ca(2+)](i) for ATP exocytosis was found to be approximately 350 nM. Subthreshold [Ca(2+)](i) transients predominantly induced vesicle docking at plasma membrane without subsequent release. ATP exocytosis triggered either by purinergic stimulation or by Ca(2+) uncaging occurred after a substantial delay ranging from tens to hundreds of seconds, with only approximately 4% of release occurring during the first 30 s. The time course of the cargo release from vesicles had two peaks centered on <or=10 s and 60 s. These results demonstrate that: (1) [Ca(2+)](i) elevations in cultured astrocytes trigger docking and release of ATP-containing vesicles; (2) vesicle docking and release have different Ca(2+) thresholds; (3) ATP exocytosis is delayed by several minutes and highly asynchronous; (4) two populations of ATP-containing vesicles with distinct (fast and slow) time course of cargo release exist in cultured astrocytes.
Collapse
Affiliation(s)
- Evgeny Pryazhnikov
- Neuroscience Center, University of Helsinki, P.O. Box 56 (Viikinkaari 4), FIN-00014, Helsinki, Finland
| | | |
Collapse
|
39
|
Porras OH, Ruminot I, Loaiza A, Barros LF. Na(+)-Ca(2+) cosignaling in the stimulation of the glucose transporter GLUT1 in cultured astrocytes. Glia 2008; 56:59-68. [PMID: 17924581 DOI: 10.1002/glia.20589] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glutamate triggers an acute stimulation of the glucose transporter GLUT1 in cultured astrocytes, a phenomenon thought to facilitate energy delivery to active areas in the brain. Here we have explored the cell signaling mechanisms involved in this response. Half-stimulation of GLUT1 occurred at low micromolar glutamate, thus within the physiological range estimated in brain interstitium. The effect was mimicked by D-aspartate and inhibited by L-threo-beta-benzyloxyaspartate or Na(+) replacement with NMDG(+), showing the participation of the Na(+)-glutamate co-transporter. AMPA and the mGLURI agonist DHPG had no effect. The stimulation of GLUT1 was fully inhibited by ouabain, but independent activation of the Na(+)/K(+) ATPase pump with gramicidin did not affect glucose transport. Simultaneous with the Na(+) rise, glutamate and D-aspartate triggered a Ca(2+)signal, whose inhibition with BAPTA prevented the stimulation of GLUT1. However, an isolated Ca(2+) signal, triggered with endothelin 1, ATP or DHPG, did not affect glucose transport. The stimulation of GLUT1 could finally be mimicked by simultaneous induction of Na(+) and Ca(2+) signals. The requirement for both cations in the stimulation of the astrocytic glucose transporter, may help to explain how glucose metabolism in the brain is strongly activated by glutamate, but not by GABA or by inter-astrocytic signaling.
Collapse
Affiliation(s)
- Omar H Porras
- Centro de Estudios Científicos (CECS), Av. Arturo Prat 514, Casilla 1469, Valdivia, Chile
| | | | | | | |
Collapse
|
40
|
Abstract
Many neurodegenerative diseases share common underlying features, most prominent of which are dysregulation of calcium homeostasis and reactive astrogliosis, ultimately triggered by oxidative stress. Interestingly, an additional feature of the early response to stress conditions is the upregulation and release of acetylcholinesterase (AChE). This study therefore investigates the link between oxidative stress, calcium influx, gene expression, protein synthesis, and AChE release. We report that, in astroglia and in an immortalized cell line, GH4-halpha7, acute oxidative stress causes influx of extracellular calcium through L-type voltage-gated calcium channels (L-VGCC), followed by increased release of AChE into the extracellular medium. Moreover, rapid and sustained changes in mRNA expression of AChE, L-VGCC, and melastatin-like transient receptor potential 2 (TRPM2) accompany profound suppression of global protein synthesis. Application of L-VGCC blockers selectively reduces stress-induced calcium influx and AChE release, mitigates changes in gene expression, and facilitates recovery from protein synthesis suppression. Although glia exhibit greater sensitivity in their responses, the results are comparable in astroglia and GH4-halpha7 cells, and suggest a generalized and integrated cellular response to stress conditions that characterizes changes observed in neurodegeneration.
Collapse
Affiliation(s)
- Cherie E Bond
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, United Kingdom.
| | | |
Collapse
|